EP2408758B1 - Crystalline tripeptide epoxy ketone protease inhibitors - Google Patents

Crystalline tripeptide epoxy ketone protease inhibitors Download PDF

Info

Publication number
EP2408758B1
EP2408758B1 EP10754225.0A EP10754225A EP2408758B1 EP 2408758 B1 EP2408758 B1 EP 2408758B1 EP 10754225 A EP10754225 A EP 10754225A EP 2408758 B1 EP2408758 B1 EP 2408758B1
Authority
EP
European Patent Office
Prior art keywords
compound
crystalline compound
proteasome
cell
treating
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP10754225.0A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP2408758A1 (en
EP2408758A4 (en
Inventor
Pasit Phiasivongsa
Louis C. Sehl
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Onyx Therapeutics Inc
Original Assignee
Onyx Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=42738216&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP2408758(B1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to EP14178725.9A priority Critical patent/EP2813241B1/en
Priority to SI201030833T priority patent/SI2408758T1/sl
Priority to PL10754225T priority patent/PL2408758T3/pl
Priority to DK14178725.9T priority patent/DK2813241T3/en
Application filed by Onyx Therapeutics Inc filed Critical Onyx Therapeutics Inc
Publication of EP2408758A1 publication Critical patent/EP2408758A1/en
Publication of EP2408758A4 publication Critical patent/EP2408758A4/en
Publication of EP2408758B1 publication Critical patent/EP2408758B1/en
Application granted granted Critical
Priority to HRP20150014AT priority patent/HRP20150014T1/hr
Priority to CY20151100026T priority patent/CY1115982T1/el
Priority to CY20161101307T priority patent/CY1118359T1/el
Priority to SM201700068T priority patent/SMT201700068B/it
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/56Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/02General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length in solution
    • C07K1/026General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length in solution by fragment condensation in solution
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0821Tripeptides with the first amino acid being heterocyclic, e.g. His, Pro, Trp
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • proteasome-mediated degradation In eukaryotes, protein degradation is predominately mediated through the ubiquitin pathway in which proteins targeted for destruction are ligated to the 76 amino acid polypeptide ubiquitin. Once targeted, ubiquitinated proteins then serve as substrates for the 26S proteasome, a multicatalytic protease, which cleaves proteins into short peptides through the action of its three major proteolytic activities. While having a general function in intracellular protein turnover, proteasome-mediated degradation also plays a key role in many processes such as major histocompatibility complex (MHC) class I antigen presentation, apoptosis, cell growth regulation, NF- ⁇ B activation, antigen processing, and transduction of pro-inflammatory signals.
  • MHC major histocompatibility complex
  • the 20S proteasome is a 700 kDa cylindrical-shaped multicatalytic protease complex comprised of 28 subunits organized into four rings. In yeast and other cukaryotes, 7 different a subunits form the outer rings and 7 different P subunits comprise the inner rings. The a subunits serve as binding sites for the 19S (PA700) and 11S (PA28) regulatory complexes, as well as a physical barrier for the inner proteolytic chamber formed by the two ⁇ subunit rings. Thus, in vivo, the proteasome is believed to exist as a 26S particle ("the 26S proteasome"). In vivo experiments have shown that inhibition of the 20S form of the proteasome can be readily correlated to inhibition of 26S proteasome.
  • N-terminal nucleophile hydrolases where the nucleophilic N-terminal residue is, for example, Cys, Ser, Thr, and other nucleophilic moieties.
  • This family includes, for example, penicillin G acylase (PGA), penicillin V acylase (PVA), glutamine PRPP amidotransferase (GAT), and bacterial glycosylasparaginase.
  • proteolytic activities have been defined for the eukaryote 20S proteasome: chymotrypsin-like activity (CT-L), which cleaves after large hydrophobic residues; trypsin-like activity (T-L), which cleaves after basic residues; and peptidylglutamyl peptide hydrolyzing activity (PGPH), which cleaves after acidic residues.
  • C-L chymotrypsin-like activity
  • T-L trypsin-like activity
  • PGPH peptidylglutamyl peptide hydrolyzing activity
  • Two additional less characterized activities have also been ascribed to the proteasome: BrAAP activity, which cleaves after branched-chain amino acids; and SNAAP activity, which cleaves after small neutral amino acids.
  • the major proteasome proteolytic activities appear to be contributed by different catalytic sites, since inhibitors, point mutations in ⁇ subunits and the exchange of ⁇ interferon-inducing ⁇ subunits
  • WO 2008/140782 discloses proteasome inhibitors.
  • One aspect of the invention relates to crystalline compounds as defined in the claims.
  • the invention relates to a crystalline compound of Formula (II)
  • Such compounds can be prepared by a method for the preparation of a crystalline compound of Formula (II), comprising one or more of: (i) preparing the amorphous compound, e.g., according to U.S. Patent Application No. 11/595,804 ; (ii) dissolving the amorphous compound in an organic solvent; (iii) bringing the solution to supersaturation; (iv) isolating the crystals, e.g., by filtering the crystals, by decanting fluid from the crystals, or by any other suitable separation technique; and (v) washing the crystals.
  • preparation further comprises inducing crystallization.
  • preparation further comprises drying, preferably under reduced pressure, such as under vacuum pressure.
  • the amorphous compound may be dissolved in a solvent selected from acetonitrile, ethyl acetate, heptanes, hexanes, isopropyl acetate, methanol, methylethyl ketone, tetrahydrofuran, toluene, and water, or any combination thereof.
  • the amorphous compound of Formula (II) may be dissolved in an organic solvent selected from acetonitrile, heptanes, hexanes, methanol, tetrahydrofuran, and toluene, or any combination thereof.
  • the organic solvent is toluene, tetrahydrofuran, or acetonitrile, preferably acetonitrile or toluene.
  • bringing the solution to supersaturation comprises the slow addition of an anti-solvent, such as water, heptanes, hexanes or another polar or nonpolar liquid miscible with the organic solvent, allowing the solution to cool (with or without seeding the solution), reducing the volume of the solution, or any combination thereof.
  • bringing the solution to supersaturation comprises adding an anti-solvent, cooling the solution to ambient temperature or lower, and reducing the volume of the solution, e.g., by evaporating solvent from the solution.
  • allowing the solution to cool may be passive (e.g., allowing the solution to stand at ambient temperature) or active (e.g., cooling the solution in an ice bath or freezer).
  • the method further comprises inducing precipitation or crystallization.
  • inducing precipitation or crystallization comprises secondary nucleation, wherein nucleation occurs in the presence of seed crystals or interactions with the environment (crystallizer walls, stirring impellers, sonication, etc.).
  • washing the crystals comprises washing with a liquid selected from anti-solvent, acetonitrile, heptanes, hexanes, methanol, tetrahydrofuran, toluene, water, or a combination thereof.
  • the crystals are washed with a combination of anti-solvent and the organic solvent.
  • the anti-solvent is water, while in other embodiments it is an alkane solvent, such as hexane or pentane, or an aromatic hydrocarbon solvent, such as benzene, toluene, or xylene.
  • washing the crystals comprises washing the crystalline compound of Formula (II) with a mix of tetrahydrofuran and an alkane solvent, such as hexanes or heptanes, or with a mix of acetonitrile and water.
  • washing the crystals comprises washing the crystalline compound of Formula (II) with toluene. In preferred such embodiments, the toluene is cooled prior to washing.
  • a crystalline compound of Formula (II) is substantially pure.
  • the melting point of the crystalline compound of Formula (II) is in the range of about 135 to about 160 °C, about 140 to about 155 °C, about 145 to about 150 °C, or even about 147 to about 149 °C, e.g., about 149 °C.
  • the DSC of a crystalline compound of Formula (II) has a sharp endothermic maximum at about 147 °C, e.g., resulting from melting and decomposition of the crystalline form as shown in Figure 1 .
  • the X-ray powder pattern of a crystalline compound of Formula (II) is ( ⁇ -2 ⁇ °): 8.94; 9.39; 9.76; 10.60; 11.09; 12.74; 15.27; 17.74; 18.96; 20.58; 20.88; 21.58; 21.78; 22.25; 22.80; 24.25; 24.66; 26.04; 26.44; 28.32; 28.96; 29.65; 30.22; 30.46; 30.78; 32.17; 33.65; 34.49; 35.08; 35.33; 37.85; 38.48 as shown in Figure 2 .
  • the TG thermogram of a crystalline compound of Formula (II) exhibits from 0.0 to 0.3% weight loss in the temperature range of 25 to 125 °C as shown in Figure 3 .
  • a crystalline compound of Formula (II) is not solvated (e.g., the crystal lattice does not comprise molecules of a solvent). In certain alternative embodiments, a crystalline compound of Formula (II) is solvated.
  • the compounds can be prepared by a method for the preparation of a crystalline compound of Formula (II), comprising (i) reacting a compound of Formula (III) wherein X is any suitable counterion, with a compound of Formula (IV) in an organic solvent (ii) preparing a solution of a compound of Formula (II) in the organic solvent; (iii) bringing the solution to supersaturation to permit formation of crystals; and (iv) isolating the crystals to provide a crystalline compound of Formula (II), e.g., by filtering the crystals, by decanting, or by any other suitable separation technique.
  • a compound of Formula (II) is not purified by chromatography prior to preparation of the solution in the organic solvent.
  • preparation further comprises inducing crystallization. In certain embodiments, preparation further comprises washing the crystals, e.g., with a solvent or non-solvent fluid. In certain embodiments, preparation further comprises drying, preferably under reduced pressure, such as under vacuum pressure.
  • X is a counterion selected from hydrobromide, hydrochloride, sulfate, phosphate, nitrate, acetate, trifluoroacetate, citrate, methanesulfonate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, succinate, tosylate, malonate, maleate, fumarate, succinate, tartrate, mesylate, 2-hydroxyethanesulfonate, and the like.
  • a counterion selected from hydrobromide, hydrochloride, sulfate, phosphate, nitrate, acetate, trifluoroacetate, citrate, methanesulfonate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, succinate, tosylate, malonate, maleate, fumarate, succinate, tartrate, mesylate, 2-hydroxyethanesulf
  • X is selected from trifluoroacetate, methanesulfonate, toluenesulfonate, acetate, chloride, and bromide, preferably trifluoroacetate.
  • the organic solvent is selected from acetonitrile, ethyl acetate, heptanes, hexanes, isopropyl acetate, methanol, methylethyl ketone, tetrahydrofuran, toluene, and water, or any combination thereof.
  • the amorphous compound of Formula (II) may be dissolved in an organic solvent selected from acetonitrile, heptanes, hexanes, methanol, tetrahydrofuran, and toluene, or any combination thereof.
  • the organic solvent is toluene, tetrahydrofuran, or acetonitrile, preferably acetonitrile or toluene.
  • preparation further comprises washing the crystals of Formula (II).
  • washing the crystals comprises washing with a liquid selected from anti-solvent, acetonitrile, heptanes, hexanes, methanol, tetrahydrofuran, toluene, water, or a combination thereof.
  • the crystals are washed with a combination of anti-solvent and the organic solvent.
  • the anti-solvent is water, while in other embodiments it is an alkane solvent, such as hexane or pentane, or an aromatic hydrocarbon solvent, such as benzene, toluene, or xylene.
  • preparation further comprises drying the crystals of both of Formula (II), preferably under reduced pressure, such as under vacuum pressure.
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a crystalline compound of Formula (II) and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition is selected from tablets, capsules, and injections.
  • proteasome Orderly protein degradation is crucial to the maintenance of normal cell functions, and the proteasome is integral to the protein degradation process.
  • the proteasome controls the levels of proteins that are important for cell-cycle progression and apoptosis in normal and malignant cells; for example, cyclins, caspases, BCL2 and nF-kB ( Kumatori et al., Proc. Natl. Acad. Sci. USA (1990) 87:7071-7075 ; Almond et al., Leukemia (2002) 16: 433-443 ).
  • inhibiting proteasome activity can translate into therapies to treat various disease states, such as malignant, non-malignant and autoimmune diseases, depending on the cells involved.
  • proteasome inhibition has already been validated as a therapeutic strategy for the treatment of multiple myeloma. This could be due, in part, to the highly proliferative malignant cell's dependency on the proteasome system to rapidly remove proteins ( Rolfe et al., J. Mol. Med. (1997) 75:5-17 ; Adams, Nature (2004) 4: 349-360 ). Therefore, certain embodiments of the invention relate to a method of treating a cancer, comprising administering to a subject in need of such treatment an effective amount of a proteasome inhibitor compound disclosed herein.
  • cancer includes, but is not limited to, blood borne and solid tumors.
  • Cancer refers to disease of blood, bone, organs, skin tissue and the vascular system, including, but not limited to, cancers of the bladder, blood, bone, brain, breast, cervix, chest, colon, endometrium, esophagus, eye, head, kidney, liver, lung, lymph nodes, mouth, neck, ovaries, pancreas, prostate, rectum, renal, skin, stomach, testis, throat, and uterus.
  • Specific cancers include, but are not limited to, leukemia (acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), hairy cell leukemia), mature B cell neoplasms (small lymphocytic lymphoma, B cell prolymphocytic leukemia, lymphoplasmacytic lymphoma (such as Waldenström's macroglobulinemia), splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, monoclonal immunoglobulin deposition diseases, heavy chain diseases, extranodal marginal zone B cell lymphoma (MALT lymphoma), nodal marginal zone B cell lymphoma (NMZL), follicular lymphoma, mantle cell lymphoma, diffuse B cell lymphoma, mediastinal (thymic) large B cell lymphoma, intravascular large
  • CMPDs chronic myeloproliferative diseases
  • the chronic myeloproliferative diseases are clonal haematopoietic stem cell disorders characterized by proliferation in the bone marrow of one or more of the myeloid lineages, resulting in increased numbers of granulocytes, red blood cells and/or platelets in the peripheral blood.
  • proteasome inhibitors for the treatment of such diseases is attractive and being examined ( Cilloni et al., Haematologica (2007) 92: 1124-1229 ).
  • CMPD can include chronic myelogenous leukaemia, chronic neutrophilic leukaemia, chronic eosinophilic leukaemia, polycythaemia vera, chronic idiopathic myelofibrosis, essential thrombocythaemia and unclassifiable chronic myeloproliferative disease.
  • An aspect of the invention is the method of treating CMPD comprising administering to a subject in need of such treatment an effective amount of a proteasome inhibitor compound disclosed herein.
  • Myelodisplastic/myeloproliferative diseases such as chronic myelomonocytic leukaemia, atypical chronic myeloid leukemia, juvenile myelomonocytic leukaemia and unclassifiable myelodysplastic/myeloproliferative disease, are characterized by hypercellularity of the bone marrow due to proliferation in one or more of the myeloid lineages. Inhibiting the proteasome with a compound or composition as described herein can serve to treat these myelodysplatic/myeloproliferative diseases by providing a subject in need of such treatment an effective amount of the compound or composition.
  • Myelodysplastic syndromes refer to a group of hematopoietic stem cell disorders characterized by dysplasia and ineffective haematopoiesis in one or more of the major myeloid cell lines.
  • Targeting NF-kB with a proteasome inhibitor in these hematologic malignancies induces apoptosis, thereby killing the malignant cell ( Braun et al. Cell Death and Differentiation (2006) 13:748-758 ).
  • a further embodiment of the invention is a method to treat MDS comprising administering to a subject in need of such treatment an effective amount of a compound disclosed herein.
  • MDS includes refractory anemia, refractory anemia with ringed sideroblasts, refractory cytopenia with multilineage dysplasia, refractory anemia with excess blasts, unclassifiable myelodysplastic syndrome and myelodysplastic syndrome associated with isolated del(5q) chromosome abnormality.
  • Mastocytosis is a proliferation of mast cells and their subsequent accumulation in one or more organ systems.
  • Mastocytosis includes, but is not limited to, cutaneous mastocytosis, indolent systemic mastocytosis (ISM), systemic mastocytosis with associated clonal haematological non-mast-cell-lineage disease (SM-AHNMD), aggressive systemic mastocytosis (ASM), mast cell leukemia (MCL), mast cell sarcoma (MCS) and extracutaneous mastocytoma.
  • Another embodiment of the invention is a method to treat mastocytosis, comprising administering an effective amount of a compound or composition disclosed herein to a subject diagnosed with mastocytosis.
  • NF- ⁇ B The proteasome regulates NF- ⁇ B, which in turn regulates genes involved in the immune and inflammatory response.
  • NF- ⁇ B is required for the expression of the immunoglobulin light chain ⁇ gene, the IL-2 receptor ⁇ -chain gene, the class I major histocompatibility complex gene, and a number of cytokine genes encoding, for example, IL-2, IL-6, granulocyte colony-stimulating factor, and IFN- ⁇ ( Palombella et al., Cell (1994) 78:773-785 ).
  • the invention relates to methods of affecting the level of expression of IL-2, MHC-I, IL-6, TNF ⁇ , IFN- ⁇ or any of the other previously-mentioned proteins, each method comprising administering to a subject an effective amount of a proteasome inhibitor compound or composition disclosed herein.
  • the invention includes a method of treating an autoimmune disease in a mammal comprising administering a therapeutically effective amount of a compound or composition described herein.
  • An "autoimmune disease” herein is a disease or disorder arising from and directed against an individual's own tissues.
  • autoimmune diseases or disorders include, but are not limited to, inflammatory responses such as inflammatory skin diseases including psoriasis and dermatitis (e.g., atopic dermatitis); systemic scleroderma and sclerosis; responses associated with inflammatory bowel disease (such as Crohn's disease and ulcerative colitis); respiratory distress syndrome (including adult respiratory distress syndrome; ARDS); dermatitis; meningitis; encephalitis; uveitis; colitis; glomerulonephritis; allergic conditions such as eczema and asthma and other conditions involving infiltration of T cells and chronic inflammatory responses; atherosclerosis; leukocyte adhesion deficiency; rheumatoid arthritis; systemic lupus erythematosus (SLE); diabetes mellitus (e.g., Type I diabetes mellitus or insulin dependent diabetes mellitis); multiple sclerosis; Reynaud's syndrome; autoimmune thyroiditis; allergic encephalo
  • the invention relates to a method of using the compound as an immunomodulatory agent for inhibiting or altering antigen presentation in a cell, comprising exposing the cell (or administering to a subject) to a compound described herein.
  • a method of treating graft or transplant-related diseases such as graft-versus-host disease or host versus-graft disease in a mammal, comprising administering a therapeutically effective amount of a compound described herein.
  • graft refers to biological material derived from a donor for transplantation into a recipient. Grafts include such diverse material as, for example, isolated cells such as islet cells; tissue such as the amniotic membrane of a newborn, bone marrow, hematopoietic precursor cells, and ocular tissue, such as corneal tissue; and organs such as skin, heart, liver, spleen, pancreas, thyroid lobe, lung, kidney, tubular organs (e.g., intestine, blood vessels, or esophagus). The tubular organs can be used to replace damaged portions of esophagus, blood vessels, or bile duct.
  • isolated cells such as islet cells
  • tissue such as the amniotic membrane of a newborn, bone marrow, hematopoietic precursor cells, and ocular tissue, such as corneal tissue
  • organs such as skin, heart, liver, spleen, pancreas, thyroid lobe, lung, kidney, tubular organs
  • the skin grafts can be used not only for bums, but also as a dressing to damaged intestine or to close certain defects such as diaphragmatic hernia.
  • the graft is derived from any mammalian source, including human, whether from cadavers or living donors. In some cases, the donor and recipient is the same mammal.
  • the graft is bone marrow or an organ such as heart and the donor of the graft and the host are matched for HLA class II antigens.
  • Histiocytic and dendritic cell neoplasms are derived from phagocytes and accessory cells, which have major roles in the processing and presentation of antigens to lymphocytes. Depleting the proteasome content in dendritic cells has been shown to alter their antigen-induced responses ( Chapatte et al. Cancer Res. (2006) 66:5461-5468 ).
  • another embodiment of the invention comprises administering an effective amount of a compound or composition disclosed herein to a subject with histiocytic or dendritic cell neoplasm.
  • Histiocytic and dendritic cell neoplasms include histiocytic sarcoma, Langerhans cell histiocytosis, Langerhans cell sarcoma, interdigitating dendritic cell sarcoma/tumor, follicular dendritic cell sarcoma/tumor and non-specified dendritic cell sarcoma.
  • an embodiment of the invention includes the treatment of lymphoproliferative diseases (LPD) associated with primary immune disorders (PID) comprising administering an effective amount of the disclosed compound to a subject in need thereof.
  • LPD lymphoproliferative diseases
  • PID primary immune disorders
  • lymphoproliferative disorders including B-cell and T-cell neoplasms and lymphomas
  • primary immunodeficiency syndromes and other primary immune disorders infection with the human immunodeficiency virus (HIV), iatrogenic immunosuppression in patients who have received solid organ or bone marrow allografts, and iatrogenic immunosuppression associated with methotrexate treatment.
  • HIV human immunodeficiency virus
  • PIDs commonly associated with LPDs are ataxia telangiectasia (AT), Wiskott-Aldrich syndrome (WAS), common variable immunodeficiency (CVID), severe combined immunodeficiency (SCID), X-linked lymphoproliferative disorder (XLP), Nijmegen breakage syndrome (NBS), hyper-IgM syndrome, and autoimmune lymphoproliferative syndrome (ALPS).
  • AT ataxia telangiectasia
  • WAS Wiskott-Aldrich syndrome
  • CVID common variable immunodeficiency
  • SCID severe combined immunodeficiency
  • XLP X-linked lymphoproliferative disorder
  • NBS Nijmegen breakage syndrome
  • hyper-IgM syndrome and autoimmune lymphoproliferative syndrome
  • Additional embodiments of the invention relate to methods for affecting the proteasome-dependent regulation of oncoproteins and methods of treating or inhibiting cancer growth, each method comprising exposing a cell (in vivo, e.g., in a subject, or in vitro ) to the proteasome inhibitor composition disclosed herein.
  • HPV-16 and HPV-18-derived E6 proteins stimulate ATP-and ubiquitin-dependent conjugation and degradation of p53 in crude reticulocyte lysates.
  • the recessive oncogene p53 has been shown to accumulate at the nonpermissive temperature in a cell line with a mutated thermolabile E1. Elevated levels of p53 may lead to apoptosis.
  • the invention relates to a method for treating p53-related apoptosis, comprising administering to a subject an effective amount of a proteasome inhibitor composition disclosed herein.
  • Another aspect of the invention relates to the use of proteasome inhibitor compositions disclosed herein for the treatment of neurodegenerative diseases and conditions, including, but not limited to, stroke, ischemic damage to the nervous system, neural trauma (e.g., percussive brain damage, spinal cord injury, and traumatic damage to the nervous system), multiple sclerosis and other immune-mediated neuropathies (e.g., Guillain-Barre syndrome and its variants, acute motor axonal neuropathy, acute inflammatory demyelinating polyneuropathy, and Fisher Syndrome), HIV/AIDS dementia complex, axonomy, diabetic neuropathy, Parkinson's disease, Huntington's disease, multiple sclerosis, bacterial, parasitic, fungal, and viral meningitis, encephalitis, vascular dementia, multi-infarct dementia, Lewy body dementia, frontal lobe dementia such as Pick's disease, subcortical dementias (such as Huntington or progressive supranuclear palsy), focal cortical atrophy syndromes (such as
  • Alzheimer's disease is characterized by extracellular deposits of ⁇ -amyloid protein ( ⁇ -AP) in senile plaques and cerebral vessels.
  • ⁇ -AP is a peptide fragment of 39 to 42 amino acids derived from an amyloid protein precursor (APP). At least three isoforms of APP are known (695, 751, and 770 amino acids). Alternative splicing of mRNA generates the isoforms; normal processing affects a portion of the ⁇ -AP sequence, thereby preventing the generation of ⁇ -AP. It is believed that abnormal protein processing by the proteasome contributes to the abundance of ⁇ -AP in the Alzheimer brain.
  • the APP-processing enzyme in rats contains about ten different subunits (22 kDa-32 kDa).
  • the 25 kDa subunit has an N-terminal sequence of X-Gln-Asn-Pro-Met-X-Thr-Gly-Thr-Ser, which is identical to the ⁇ -subunit of human macropain ( Kojima, S. et al., Fed. Eur. Biochem. Soc., (1992) 304:57-60 ).
  • the APP-processing enzyme cleaves at the Gln 15 -Lys 16 bond; in the presence of calcium ion, the enzyme also cleaves at the Met -1 --Asp 1 bond and the Asp 1 --Ala 2 bond to release the extracellular domain of ⁇ -AP.
  • One aspect of the invention therefore, relates to a method of treating Alzheimer's disease, comprising administering to a subject an effective amount of a proteasome inhibitor compound or composition disclosed herein.
  • Such treatment includes reducing the rate of ⁇ -AP processing, reducing the rate of ⁇ -AP plaque formation, reducing the rate of ⁇ -AP generation, and reducing the clinical signs of Alzheimer's disease.
  • Fibrosis is the excessive and persistent formation of fibrous connective tissue resulting from the hyperproliferative growth of fibroblasts and is associated with activation of the TGF- ⁇ signaling pathway. Fibrosis involves extensive deposition of extracellular matrix and can occur within virtually any tissue or across several different tissues. Normally, the level of intracellular signaling protein (Smad) that activates transcription of target genes upon TGF- ⁇ stimulation is regulated by proteasome activity (Xu et al., 2000).
  • Smad intracellular signaling protein
  • fibrotic conditions such as cystic fibrosis, injection fibrosis, endomyocardial fibrosis, idiopathic pulmonary fibrosis, myelofibrosis, retroperitoneal fibrosis, progressive massive fibrosis, nephrogenic systemic fibrosis.
  • Other conditions that are often associated with fibrosis include cirrhosis, diffuse parenchymal lung disease, post-vasectomy pain syndrome, tuberculosis, sickle-cell anemia and rheumatoid arthritis.
  • An embodiment of the invention is the method of treating a fibrotic or fibrotic-associated condition comprising administering an effective amount of the composition described herein to a subject in need of such treatment.
  • the invention relates to the topical or systemic administration of a subject inhibitor to treat burns. Wound closure following surgery is often associated with disfiguring scars, which may be prevented by inhibition of fibrosis. Thus, in certain embodiments, the invention relates to a method for the prevention or reduction of scarring.
  • LPS lipopolysaccharide
  • TNF ⁇ lipopolysaccharide
  • the first step in the activation of cells by LPS is the binding of LPS to specific membrane receptors.
  • the ⁇ - and ⁇ -subunits of the 20S proteasome complex have been identified as LPS-binding proteins, suggesting that the LPS-induced signal transduction may be an important therapeutic target in the treatment or prevention of sepsis ( Qureshi, N. et al., J. Immun. (2003) 171: 1515-1525 ). Therefore, in certain embodiments, the proteasome inhibitor composition may be used for the inhibition of TNF ⁇ to prevent and/or treat septic shock.
  • Ischemia and reperfusion injury results in hypoxia, a condition in which there is a deficiency of oxygen reaching the tissues of the body. This condition causes increased degradation of I ⁇ -B ⁇ , thereby resulting in the activation of NF- ⁇ B (Koong et al., 1994). It has been demonstrated that the severity of injury resulting in hypoxia can be reduced with the administration of a proteasome inhibitor (Gao et al., 2000; Bao et al., 2001; Pye et al., 2003). Therefore, certain embodiments of the invention relate to a method of treating an ischemic condition or reperfusion injury comprising administering to a subject in need of such treatment an effective amount of the proteasome inhibitor compound disclosed herein.
  • Such conditions or injuries include, but are not limited to, acute coronary syndrome (vulnerable plaques), arterial occlusive disease (cardiac, cerebral, peripheral arterial and vascular occlusions), atherosclerosis (coronary sclerosis, coronary artery disease), infarctions, heart failure, pancreatitis, myocardial hypertrophy, stenosis, and restenosis.
  • acute coronary syndrome vulnerable plaques
  • arterial occlusive disease cardiac, cerebral, peripheral arterial and vascular occlusions
  • atherosclerosis coronary sclerosis, coronary artery disease
  • infarctions heart failure
  • pancreatitis myocardial hypertrophy
  • stenosis stenosis
  • restenosis examples of such conditions or injuries
  • NF- ⁇ B also binds specifically to the HIV-enhancer/promoter.
  • the HIV regulatory protein Nef of pbj14 differs by two amino acids in the region which controls protein kinase binding. It is believed that the protein kinase signals the phosphorylation of I ⁇ B, triggering I ⁇ B degradation through the ubiquitin-proteasome pathway. After degradation, NF- ⁇ B is released into the nucleus, thus enhancing the transcription of HIV ( Cohen, J., Science, (1995) 267:960 ).
  • the invention relates to a method for inhibiting or reducing HIV infection in a subject, or a method for decreasing the level of viral gene expression, each method comprising administering to the subject an effective amount of a proteasome inhibitor compound or composition disclosed herein.
  • Viral infections contribute to the pathology of many diseases.
  • Heart conditions such as ongoing myocarditis and dilated cardiomyopathy have been linked to the coxsackievirus B3.
  • specific proteasome subunits were uniformly up-regulated in hearts of mice which developed chronic myocarditis ( Szalay et al, Am J Pathol 168:1542-52, 2006 ).
  • Some viruses utilize the ubiquitin-proteasome system in the viral entry step where the virus is released from the endosome into the cytosol.
  • the mouse hepatitis virus (MHV) belongs to the Coronaviridae family, which also includes the severe acute respiratory syndrome (SARS) coronavirus.
  • the invention relates to a method for treating viral infection, such as SARS or hepatitis A, B, C, D and E, comprising contacting a cell with (or administering to a subject) an effective amount of a compound or composition disclosed herein.
  • the disclosed compositions may be useful for the treatment of a parasitic infection, such as infections caused by protozoan parasites.
  • a parasitic infection such as infections caused by protozoan parasites.
  • the proteasome of these parasites is considered to be involved primarily in cell differentiation and replication activities ( Paugam et al., Trends Parasitol. 2003, 19(2): 55-59 ).
  • entamoeba species have been shown to lose encystation capacity when exposed to proteasome inhibitors ( Gonzales, et al., Arch. Med. Res. 1997, 28, Spec No: 139-140 ).
  • the administrative protocols for the proteasome inhibitor compositions are useful for the treatment of parasitic infections in humans caused by a protozoan parasite selected from Plasmodium sps.
  • Trypanosoma sps. including T. cruzi, which causes Chagas' disease, and T. brucei which causes African sleeping sickness
  • Leishmania sps. including L. amazonesis, L. donovani, L. infantum, L. mexicana, etc.
  • Pneumocystis carinii a protozoan known to cause pneumonia in AIDS and other immunosuppressed patients
  • Toxoplasma gondii Entamoeba histolytica, Entamoeba invadens, and Giardia lamblia.
  • the disclosed proteasome inhibitor compositions are useful for the treatment of parasitic infections in animals and livestock caused by a protozoan parasite selected from Plasmodium hermani, Cryptosporidium sps., Echinococcus granulosus, Eimeria tenella, Sarcocystis neurona, and Neurospora crassa.
  • a protozoan parasite selected from Plasmodium hermani, Cryptosporidium sps., Echinococcus granulosus, Eimeria tenella, Sarcocystis neurona, and Neurospora crassa.
  • Other compounds that act as proteasome inhibitors in the treatment of parasitic diseases are described in WO 98/10779 .
  • the proteasome inhibitor compositions inhibit proteasome activity in a parasite without recovery in red blood cells and white blood cells.
  • the long half-life of blood cells may provide prolonged protection with regard to therapy against recurring exposures to parasites.
  • the proteasome inhibitor compositions may provide prolonged protection with regard to chemoprophylaxis against future infection.
  • Prokaryotes have an equivalent to the eukaryote 20S proteasome particle. Although the subunit composition of the prokaryote 20S particle is simpler than that of eukaryotes, it has the ability to hydrolyze peptide bonds in a similar manner. For example, the nucleophilic attack on the peptide bond occurs through the threonine residue on the N-terminus of the ⁇ -subunits.
  • an embodiment of this invention relates to a method of treating prokaryotic infections, comprising administering to a subject an effective amount of a proteasome inhibitor compound or composition disclosed herein.
  • Prokaryotic infections may include diseases caused by either mycobacteria (such as tuberculosis, leprosy or Buruli ulcer) or archaebacteria.
  • proteasome inhibitors that bind to the 20S proteasome stimulate bone formation in bone organ cultures. Furthermore, when such inhibitors have been administered systemically to mice, certain proteasome inhibitors increased bone volume and bone formation rates over 70% ( Garrett, I. R. et al., J. Clin. Invest. (2003) 111: 1771-1782 ), therefore suggesting that the ubiquitin-proteasome machinery regulates osteoblast differentiation and bone formation. Therefore, a disclosed proteasome inhibitor compound or composition may be useful in the treatment and/or prevention of diseases associated with bone loss, such as osteoporosis.
  • the invention relates to a method for treating a disease or condition selected from cancer, autoimmune disease, graft or transplant-related condition, neurodegenerative disease, fibrotic-associated condition, ischemic-related conditions, infection (viral, parasitic or prokaryotic) and diseases associated with bone loss, comprising administering a compound or composition as disclosed herein.
  • Compounds prepared as described herein can be administered in various forms, depending on the disorder to be treated and the age, condition, and body weight of the patient, as is well known in the art.
  • the compounds may be formulated as tablets, capsules, granules, powders, or syrups; or for parenteral administration, they may be formulated as injections (intravenous, intramuscular, or subcutaneous), drop infusion preparations, or suppositories.
  • injections intravenous, intramuscular, or subcutaneous
  • drop infusion preparations or suppositories.
  • ophthalmic mucous membrane route they may be formulated as eye drops or eye ointments.
  • the active ingredient may be mixed with any conventional additive or excipient, such as a binder, a disintegrating agent, a lubricant, a corrigent, a solubilizing agent, a suspension aid, an emulsifying agent, a coating agent, a cyclodextrin, and/or a buffer.
  • a daily dosage of from 0.01 to 2000 mg of the compound is recommended for an adult human patient, and this may be administered in a single dose or in divided doses.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect.
  • the precise time of administration and/or amount of the composition that will yield the most effective results in terms of efficacy of treatment in a given patient will depend upon the activity, pharmacokinetics, and bioavailability of a particular compound, physiological condition of the patient (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication), route of administration, etc.
  • physiological condition of the patient including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication
  • route of administration etc.
  • the above guidelines can be used as the basis for fine-tuning the treatment, e.g., determining the optimum time and/or amount of administration, which will require no more than routine experimentation consisting of monitoring the subject and adjusting the dosage and/or timing.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those ligands, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose, and sucrose; (2) starches, such as corn starch, potato starch, and substituted or unsubstituted ⁇ -cyclodextrin; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate;
  • pharmaceutically acceptable salt refers to the relatively non-toxic, inorganic and organic acid addition salts of the inhibitor(s). These salts can be prepared in situ during the final isolation and purification of the inhibitor(s), or by separately reacting a purified inhibitor(s) in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, laurylsulphonate salts, and amino acid salts, and the like.
  • sulfate bisulfate
  • phosphate nitrate
  • acetate valerate
  • oleate palmitate
  • stearate laurate
  • benzoate lactate
  • phosphate tosylate
  • citrate maleate
  • fumarate succinate
  • tartrate naphthylate
  • mesylate glucoheptonate
  • lactobionate lactobionate
  • laurylsulphonate salts
  • the inhibitors useful in the methods of the present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable bases.
  • pharmaceutically acceptable salts refers to the relatively non-toxic inorganic and organic base addition salts of an inhibitor(s). These salts can likewise be prepared in situ during the final isolation and purification of the inhibitor(s), or by separately reacting the purified inhibitor(s) in its free acid form with a suitable base, such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine.
  • Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like.
  • Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like (see, for example, Berge et al., supra).
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring, and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT
  • Formulations suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or nonaqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert matrix, such as gelatin and glycerin, or sucrose and acacia) and/or as mouthwashes, and the like, each containing a predetermined amount of an inhibitor(s) as an active ingredient.
  • a composition may also be administered as a bolus, electuary, or paste.
  • the active ingredient may be mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, cyclodextrins, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose, and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as par
  • the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols, and the like.
  • the crystalline tripeptide epoxyketone is administered to a mammal as a capsule.
  • the crystalline tripeptide epoxyketone is a compound of formula (I).
  • the crystalline tripeptide epoxyketone is a compound of formula (II).
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered inhibitor(s) moistened with an inert liquid diluent.
  • Tablets, and other solid dosage forms may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes, and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • opacifying agents include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents, and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols, and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents, and e
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.
  • Suspensions in addition to the active inhibitor(s) may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more inhibitor(s) with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams, or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of an inhibitor(s) include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, and inhalants.
  • the active component may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • the ointments, pastes, creams, and gels may contain, in addition to inhibitor(s), excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to an inhibitor(s), excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates, and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • the inhibitor(s) can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation, or solid particles containing the composition.
  • a nonaqueous (e.g., fluorocarbon propellant) suspension could be used.
  • Sonic nebulizers are preferred because they minimize exposing the agent to shear, which can result in degradation of the compound.
  • an aqueous aerosol is made by formulating an aqueous solution or suspension of the agent together with conventional pharmaceutically acceptable carriers and stabilizers.
  • the carriers and stabilizers vary with the requirements of the particular composition, but typically include nonionic surfactants (Tweens, Pluronics, sorbitan esters, lecithin, Cremophors), pharmaceutically acceptable co-solvents such as polyethylene glycol, innocuous proteins like serum albumin, oleic acid, amino acids such as glycine, buffers, salts, sugars, or sugar alcohols.
  • Aerosols generally are prepared from isotonic solutions.
  • Transdermal patches have the added advantage of providing controlled delivery of an inhibitor(s) to the body.
  • dosage forms can be made by dissolving or dispersing the agent in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the inhibitor(s) across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the inhibitor(s) in a polymer matrix or gel.
  • compositions of this invention suitable for parenteral administration comprise one or more inhibitors(s) in combination with one or more pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include tonicity-adjusting agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • adjuvants such as preservatives, wetting agents, emulsifying agents, and dispersing agents.
  • Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include tonicity-adjusting agents, such as sugars
  • delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • Injectable depot forms are made by forming microcapsule matrices of inhibitor(s) in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection, and infusion.
  • systemic administration means the administration of a ligand, drug, or other material other than directly into the central nervous system, such that it enters the patient's system and thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
  • inhibitors(s) may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally, and topically, as by powders, ointments or drops, including buccally and sublingually.
  • the inhibitor(s), which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • compositions of this invention may be provided in an aqueous solution containing about 0.1-10% w/v of a compound disclosed herein, among other substances, for parenteral administration. Typical dose ranges are from about 0.01 to about 50 mg/kg of body weight per day, given in 1-4 divided doses. Each divided dose may contain the same or different compounds of the invention.
  • the dosage will be an effective amount depending on several factors including the overall health of a patient, and the formulation and route of administration of the selected compound(s).
  • C x-y alkyl refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups that contain from x to y carbons in the chain, including haloalkyl groups such as trifluoromethyl and 2,2,2-trifluoroethyl, etc.
  • C 0 alkyl indicates a hydrogen where the group is in a terminal position, a bond if internal.
  • C 2-y alkenyl and “C 2-y alkynyl” refer to substituted or unsubstituted unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
  • alkoxy refers to an alkyl group having an oxygen attached thereto. Representative alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like.
  • An "ether” is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxy.
  • C 1-6 alkoxyalkyl refers to a C 1-6 alkyl group substituted with an alkoxy group, thereby forming an ether.
  • C 1-6 aralkyl refers to a C 1-6 alkyl group substituted with an aryl group.
  • amine and “amino” are art-recognized and refer to both unsubstituted and substituted amines and salts thereof, e.g., a moiety that can be represented by the general formulae: wherein R 9 , R 10 and R 10' each independently represent a hydrogen, an alkyl, an alkenyl, -(CH 2 ) m -R 8 , or R 9 and R 10 taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; R 8 represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocyclyl or a polycyclyl; and m is zero or an integer from 1 to 8.
  • R 9 or R 10 can be a carbonyl, e.g., R 9 , R 10 , and the nitrogen together do not form an imide.
  • R 9 and R 10 each independently represent a hydrogen, an alkyl, an alkenyl, or -(CH 2 ) m -R 8 .
  • the amino group is basic, meaning the protonated form has a pK a ⁇ 7.00.
  • amide and “amido” are art-recognized as an amino-substituted carbonyl and includes a moiety that can be represented by the general formula: wherein R 9 , R 10 are as defined above. Preferred embodiments of the amide will not include imides which may be unstable.
  • aryl as used herein includes 5-, 6-, and 7-membered substituted or unsubstituted single-ring aromatic groups in which each atom of the ring is carbon.
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline, and the like.
  • carrier refers to a non-aromatic substituted or unsubstituted ring in which each atom of the ring is carbon.
  • carrier also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is carbocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • carbonyl is art-recognized and includes such moieties as can be represented by the general formula: wherein X is a bond or represents an oxygen or a sulfur, and R 11 represents a hydrogen, an alkyl, an alkenyl, -(CH 2 ) m -R 8 or a pharmaceutically acceptable salt, R 11' represents a hydrogen, an alkyl, an alkenyl or -(CH 2 ) m -R 8 , where m and R 8 are as defined above. Where X is an oxygen and R 11 or R 11' is not hydrogen, the formula represents an "ester". Where X is an oxygen, and R 11 is a hydrogen, the formula represents a "carboxylic acid".
  • heteroaryl includes substituted or unsubstituted aromatic 5- to 7-membered ring structures, more preferably 5- to 6-membered rings, whose ring structures include one to four heteroatoms.
  • heteroaryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, isoxazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like.
  • heteroatom as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, phosphorus, and sulfur.
  • heterocyclyl or “heterocyclic group” refer to substituted or unsubstituted non-aromatic 3- to 10-membered ring structures, more preferably 3- to 7-membered rings, whose ring structures include one to four heteroatoms.
  • heterocyclyl or “heterocyclic group” also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Heterocyclyl groups include, for example, tetrahydrofuran, piperidine, piperazine, pyrrolidine, morpholine, lactones, lactams, and the like.
  • C 1-6 heterocycloalkyl refers to a C 1-6 alkyl group substituted with a heterocyclyl group.
  • C 1-6 hydroxyalkyl refers to a C 1-6 alkyl group substituted with a hydroxy group.
  • inhibitor is meant to describe a compound that blocks or reduces an activity of an enzyme (for example, inhibition of proteolytic cleavage of standard fluorogenic peptide substrates such as suc-LLVY-AMC, Box-LLR-AMC and Z-LLE-AMC, inhibition of various catalytic activities of the 20S proteasome).
  • An inhibitor can act with competitive, uncompetitive, or noncompetitive inhibition.
  • An inhibitor can bind reversibly or irreversibly, and therefore the term includes compounds that are suicide substrates of an enzyme.
  • An inhibitor can modify one or more sites on or near the active site of the enzyme, or it can cause a conformational change elsewhere on the enzyme.
  • orally bioavailable is meant to describe a compound administered to a mouse at 40 mg/kg or less, 20 mg/kg or less, or even 10 mg/kg or less, wherein one hour after oral administration such a compound shows at least about 50%, at least about 75% or even at least about 90% inhibition of proteasome CT-L activity in the blood.
  • peptide includes not only standard amide linkage with standard ⁇ -substituents, but commonly utilized peptidomimetics, other modified linkages, non-naturally occurring side chains, and side chain modifications, as detailed below.
  • polycyclyl or “polycyclic” refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are "fused rings".
  • rings e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls
  • Each of the rings of the polycycle can be substituted or unsubstituted.
  • proteasome as used herein is meant to include immuno- and constitutive proteasomes.
  • substantially pure refers to a crystalline polymorph that is greater than 90% pure, meaning that contains less than 10% of any other compound, including the corresponding amorphous compound.
  • the crystalline polymorph is greater than 95% pure, or even greater than 98% pure.
  • preventing is art-recognized, and when used in relation to a condition, such as a local recurrence (e.g., pain), a disease such as cancer, a syndrome complex such as heart failure or any other medical condition, is well understood in the art, and includes administration of a composition which reduces the frequency of, or delays the onset of, symptoms of a medical condition in a subject relative to a subject which does not receive the composition.
  • a condition such as a local recurrence (e.g., pain)
  • a disease such as cancer
  • a syndrome complex such as heart failure or any other medical condition
  • prevention of cancer includes, for example, reducing the number of detectable cancerous growths in a population of patients receiving a prophylactic treatment relative to an untreated control population, and/or delaying the appearance of detectable cancerous growths in a treated population versus an untreated control population, e.g., by a statistically and/or clinically significant amount.
  • Prevention of an infection includes, for example, reducing the number of diagnoses of the infection in a treated population versus an untreated control population, and/or delaying the onset of symptoms of the infection in a treated population versus an untreated control population.
  • Prevention of pain includes, for example, reducing the magnitude of, or alternatively delaying, pain sensations experienced by subjects in a treated population versus an untreated control population.
  • prodrug encompasses compounds that, under physiological conditions, are converted into therapeutically active agents.
  • a common method for making a prodrug is to include selected moieties that are hydrolyzed under physiological conditions to reveal the desired molecule.
  • the prodrug is converted by an enzymatic activity of the host animal.
  • prophylactic or therapeutic treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
  • the unwanted condition e.g., disease or other unwanted state of the host animal
  • proteasome as used herein is meant to include immuno- and constitutive proteasomes.
  • substituted refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
  • Substituents can include, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by
  • a “therapeutically effective amount” of a compound with respect to the subject method of treatment refers to an amount of the compound(s) in a preparation which, when administered as part of a desired dosage regimen (to a mammal, preferably a human) alleviates a symptom, ameliorates a condition, or slows the onset of disease conditions according to clinically acceptable standards for the disorder or condition to be treated or the cosmetic purpose, e.g., at a reasonable benefit/risk ratio applicable to any medical treatment.
  • thioether refers to an alkyl group, as defined above, having a sulfur moiety attached thereto.
  • the "thioether” is represented by -S-alkyl.
  • Representative thioether groups include methylthio, ethylthio, and the like.
  • treating includes reversing, reducing, or arresting the symptoms, clinical signs, and underlying pathology of a condition in manner to improve or stabilize a subject's condition.
  • N-Boc phenylalanine-ketoepoxide 140 mg, 0.46 mmol was diluted with DCM (2 mL) and cooled to 0 °C. To this solution was added trifluoroacetic acid (6 mL). The cooling bath was removed and the reaction stirred for 1 hour at which time TLC showed complete consumption of starting material. The resulting solution was concentrated under reduced pressure and placed under high vacuum to yield TFA salt of Compound (H).
  • Amorphous Compound 1 (50 mg) was dissolved in acetonitrile (1 mL), then deionized water (2 mL) was added, and the solution brought to supersaturation by slowly evaporating off 1 mL over about 1-2 weeks. The resulting crystals were filtered, washed with 1 mL 1:2 acetonitrile-water, and dried under vacuum for 12 hours to provide a crystalline polymorph of Compound 1 (25 mg) with a melting point of 148 °C.
  • the characteristic DSC curve of the sample is shown in Figure 1 as recorded on a TA Instruments Differential Scanning Calorimeter 2920 at a heating rate of 10 °C/minute.
  • Amorphous Compound 1 (611 mg) was dissolved in tetrahydrofuran (5 mL), followed by addition of hexanes (5 mL) and the solution was seeded with crystalline polymorph Compound 1 as prepared in Example 2, and the solution brought to supersaturation by slowly evaporating off 5 mL over about 17 hours. The resulting crystals were filtered, washed with 1 mL 1:1 tetrahydrofuran-hexanes, and dried under vacuum for 12 hours to provide a crystalline polymorph of Compound 1 (150 mg) with a melting point of 147 °C.
  • Amorphous Compound 1 (176 mg) was dissolved in tetrahydrofuran (5 mL), then toluene (25 mL) was added. The solution was seeded with crystalline polymorph Compound 1 as prepared in Example 2, and the solution was brought to supersaturation by slowly evaporating off 20 mL over about 2 days. The resulting crystals were filtered, washed with 15 mL toluene, and dried under vacuum for 12 hours to provide a crystalline polymorph of Compound 1 (88 mg) with a melting point of 149 °C.
  • Amorphous Compound 1 (312 mg) was dissolved in toluene (50 mL), heated to about 100 °C to complete dissolution, then hexanes (50 mL) were added and the solution was seeded with crystalline polymorph Compound 1 as prepared in Example 2, and the solution brought to supersaturation by slowly evaporating off 60 mL over about 2 days. The resulting crystals were filtered, washed with 10 mL toluene, and dried under vacuum for 12 hours to provide a crystalline polymorph of Compound 1 (156 mg) with a melting point of 149 °C.
  • Amorphous Compound 1 (1.4 g) was dissolved in toluene (25 mL), heated to about 50 °C to complete dissolution, then brought to supersaturation by cooling to 22 °C and allowing the compound to crystallize for 12 hours. The resulting crystals were filtered, washed with 5 mL hexanes, and dried under vacuum for 12 hours to provide a crystalline polymorph of Compound 1 (0.94 g) with a melting point of 149 °C.
  • N-Boc phenylalanine-ketoepoxide (1.0 equivalent) was dissolved in DCM (3 L/kg of N-Boc phenylalanine-ketoepoxide) in a 3-neck round bottom flask under inert atmosphere and the solution was cooled in ice bath. Then, TFA (5.0 equivalents) was added at a rate to maintain the internal temperature below 10 °C. The reaction mixture was then warmed to approximately 20 °C and stirred for 1 to 3 hours. MTBE (3.6 L/kg of N-Boc phenylalanine-ketoepoxide) was then added to the reaction mixture while maintaining mixture temperature below 25 °C.
  • Crude Compound 1 was precipitated by pouring the reaction mixture onto 8% sodium bicarbonate (40 L/kg of Compound (G)) and the suspension of crude Compound 1 was stirred for 12 hours at 20 to 25 °C, followed by stirring at 0 to 5 °C for 1 hour.
  • the white solid was filtered and rinsed with water (5 L/kg of Compound (G)).
  • the white solid was then reslurried in water (15 L/kg) for 3 hours at 20 to 25 °C, filtered and rinsed with water (5 L/kg of Compound (G)) and isopropyl acetate (2 x 2 L/kg of Compound (G)).
  • the white solid was dried under vacuum at 45 °C to constant weight. Yield of crude Compound 1 was 65%, with HPLC purity of 97.2%.
  • the reaction mixture was quenched by addition of pre-chilled saturated sodium bicarbonate (94 L/kg of Compound (G)), while maintaining internal temperature of less 10 °C. The content was then transferred to a separatory funnel. The mixture was extracted with ethyl acetate (24 L/kg of Compound (G)), and the organic layer was washed with saturated sodium bicarbonate (12 L/kg of Compound (G)) and with saturated sodium chloride (12 L/kg of Compound (G).
  • the organic layer was concentrated under reduced pressure with a bath temperature of less than 30 °C to 15 L/kg of Compound (G), followed by co-distillation with isopropyl acetate (2 x 24 L/kg of PR-022). Final volume was adjusted to 82 L/kg of Compound (G) with isopropyl acetate before heating to 60 °C to obtain a clear solution.
  • the clear solution mixture was allowed to cool to 50 °C before adding seed crystals.
  • the solution was allowed to cool to 20 °C, when substantial crystallization of Compound 1 had occurred.
  • the suspension was stirred at 0 °C for 12 hours before filtration and rinsing with isopropyl acetate (2 L/kg of Compound 1).
  • Compound 1 was dried under vacuum at 20 °C for 12 hours to constant weight. Yield of Compound 1 was 48%, with HPLC purity of 97.4%.
EP10754225.0A 2009-03-20 2010-03-22 Crystalline tripeptide epoxy ketone protease inhibitors Active EP2408758B1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
EP14178725.9A EP2813241B1 (en) 2009-03-20 2010-03-22 Crystalline tripeptide epoxy ketone protease inhibitors
SI201030833T SI2408758T1 (sl) 2009-03-20 2010-03-22 Kristalni tripeptidni epoksi ketonski proteazni inhibotorji
PL10754225T PL2408758T3 (pl) 2009-03-20 2010-03-22 Krystaliczne tripeptydowe epoksyketonowe inhibitory proteazy
DK14178725.9T DK2813241T3 (en) 2009-03-20 2010-03-22 Crystalline tripeptide epoxy-ketone protease inhibitors
HRP20150014AT HRP20150014T1 (hr) 2009-03-20 2015-01-08 Inhibitori proteaze kristalnih tripeptida epoksidketona
CY20151100026T CY1115982T1 (el) 2009-03-20 2015-01-12 Αναστολεις κρυσταλλικων πρωτεασων τριπεπτιδικης εποξικης κετονης
CY20161101307T CY1118359T1 (el) 2009-03-20 2016-12-16 Αναστολεις κρυσταλλικων πρωτεασων τριπεπτιδικης εποξικης κετονης
SM201700068T SMT201700068B (it) 2009-03-20 2017-01-30 Inibitori cristallini del tripeptide epossichetone proteasi

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US16219609P 2009-03-20 2009-03-20
US18056109P 2009-05-22 2009-05-22
PCT/US2010/028126 WO2010108172A1 (en) 2009-03-20 2010-03-22 Crystalline tripeptide epoxy ketone protease inhibitors

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP14178725.9A Division EP2813241B1 (en) 2009-03-20 2010-03-22 Crystalline tripeptide epoxy ketone protease inhibitors
EP14178725.9A Division-Into EP2813241B1 (en) 2009-03-20 2010-03-22 Crystalline tripeptide epoxy ketone protease inhibitors

Publications (3)

Publication Number Publication Date
EP2408758A1 EP2408758A1 (en) 2012-01-25
EP2408758A4 EP2408758A4 (en) 2012-09-05
EP2408758B1 true EP2408758B1 (en) 2014-10-22

Family

ID=42738216

Family Applications (2)

Application Number Title Priority Date Filing Date
EP10754225.0A Active EP2408758B1 (en) 2009-03-20 2010-03-22 Crystalline tripeptide epoxy ketone protease inhibitors
EP14178725.9A Active EP2813241B1 (en) 2009-03-20 2010-03-22 Crystalline tripeptide epoxy ketone protease inhibitors

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP14178725.9A Active EP2813241B1 (en) 2009-03-20 2010-03-22 Crystalline tripeptide epoxy ketone protease inhibitors

Country Status (45)

Country Link
US (5) US8604215B2 (es)
EP (2) EP2408758B1 (es)
JP (1) JP5723357B2 (es)
KR (1) KR101729344B1 (es)
CN (1) CN102428075B (es)
AP (1) AP3513A (es)
AR (1) AR075899A1 (es)
AU (1) AU2010226410B2 (es)
BR (1) BRPI1009369A2 (es)
CA (1) CA2755971C (es)
CL (1) CL2011002326A1 (es)
CO (1) CO6430433A2 (es)
CR (1) CR20110491A (es)
CU (1) CU20110176A7 (es)
CY (1) CY1118359T1 (es)
DK (2) DK2408758T3 (es)
DO (1) DOP2011000286A (es)
EA (2) EA024672B1 (es)
EC (1) ECSP11011341A (es)
ES (2) ES2614557T3 (es)
GE (1) GEP20156392B (es)
HK (2) HK1162476A1 (es)
HN (1) HN2011002459A (es)
HR (2) HRP20150014T1 (es)
HU (1) HUE032430T2 (es)
IL (1) IL215174A (es)
LT (1) LT2813241T (es)
MA (1) MA33197B1 (es)
ME (1) ME01277B (es)
MX (2) MX343562B (es)
MY (1) MY156522A (es)
NI (1) NI201100170A (es)
NZ (2) NZ595847A (es)
PE (1) PE20120645A1 (es)
PL (2) PL2408758T3 (es)
PT (2) PT2408758E (es)
RS (2) RS53746B1 (es)
SA (2) SA114350283B1 (es)
SG (2) SG174446A1 (es)
SI (2) SI2813241T1 (es)
SM (2) SMT201500009B (es)
TN (1) TN2011000470A1 (es)
TW (1) TWI504598B (es)
WO (1) WO2010108172A1 (es)
ZA (2) ZA201106826B (es)

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105237621A (zh) 2005-11-09 2016-01-13 欧尼斯治疗公司 用于酶抑制的化合物
EP2041158B1 (en) 2006-06-19 2013-04-17 Onyx Therapeutics, Inc. Peptide epoxyketones for proteasome inhibition
CL2008002966A1 (es) 2007-10-04 2010-06-25 Onyx Therapeutics Inc Compuesto tetrapeptido ceto-epoxido cristalino; sal citrato cristalina del compuesto; metodos de preparacion; compuesto intermediario cristalino; metodo de preparacion; y uso para tratar cancer, enfermedad autoinmune, afeccion relacionada con trasplante, enfermedad neurodegenerativa, afeccion asociada con fibrosis, entre otros.
EA035100B1 (ru) 2008-10-21 2020-04-28 Оникс Терапьютикс, Инк. Комбинированная терапия с применением пептид эпоксикетонов
TWI504598B (zh) 2009-03-20 2015-10-21 Onyx Therapeutics Inc 結晶性三肽環氧酮蛋白酶抑制劑
PT3178321T (pt) 2009-10-07 2019-08-01 Dow Agrosciences Llc Misturas fungicidas sinérgicas de epoxiconazol para o controlo de fungos em cereais
WO2011060179A1 (en) 2009-11-13 2011-05-19 Onyx Therapeutics, Inc Use of peptide epoxyketones for metastasis suppression
US9359398B2 (en) 2010-03-01 2016-06-07 Onyx Therapeutics, Inc. Compounds for immunoproteasome inhibition
BR112012025264A2 (pt) 2010-04-07 2019-09-24 Onyx Therapeutics Inc inibidor de imunoproteassoma de e´poxicetona peptídica cristalina.
UY34897A (es) 2012-07-09 2014-01-31 Onyx Therapeutics Inc Profarmacos de inhibidores peptidicos de expoxi cetona proteasa
EP2883960B1 (en) * 2012-08-10 2020-05-13 Ajinomoto Co., Inc. METHOD FOR PRODUCING gamma-GLUTAMYL-VALYL-GLYCINE CRYSTAL
TW201422255A (zh) * 2012-10-24 2014-06-16 Onyx Therapeutics Inc 用於蛋白酶體抑制劑之調整釋放製劑
ES2666144T3 (es) 2012-12-28 2018-05-03 Dow Agrosciences Llc Mezclas fungicidas sinérgicas para control fúngico en cereales
WO2015100184A1 (en) 2013-12-26 2015-07-02 Dow Agrosciences Llc Use of macrocyclic picolinamides as fungicides
CN104945470B (zh) * 2014-03-30 2020-08-11 浙江大学 杂环构建的三肽环氧酮类化合物及制备和应用
CN104974221B (zh) * 2014-04-03 2020-10-23 中国医学科学院药物研究所 二肽及三肽类蛋白酶体抑制剂及其制法和药物用途
BR112017000169A2 (pt) 2014-07-08 2017-10-31 Dow Agrosciences Llc picolinamidas macrocíclicas como fungicidas
WO2016069479A1 (en) 2014-10-27 2016-05-06 Apicore Us Llc Methods of making carfilzomib and intermediates thereof
BR122019026066B1 (pt) 2014-12-30 2022-01-18 Dow Agrosciences Llc Compostos de picolinamida
WO2016109288A1 (en) 2014-12-30 2016-07-07 Dow Agrosciences Llc Use of picolinamide compounds with fungicidal activity
CA2972403A1 (en) 2014-12-30 2016-07-07 Dow Agrosciences Llc Picolinamide compounds with fungicidal activity
CA2972034A1 (en) 2014-12-30 2016-07-07 Dow Agrosciences Llc Picolinamides with fungicidal activity
BR112017013608B8 (pt) 2014-12-30 2022-08-23 Dow Agrosciences Llc Picolinamidas como fungicidas
CN105949279A (zh) * 2016-04-27 2016-09-21 浙江大学 蛋白酶体抑制剂Oprozomib及其类似物的制备方法
AR108919A1 (es) 2016-06-29 2018-10-10 Kezar Life Sciences Proceso de preparación del inhibidor de inmunoproteasoma de péptido epoxicetona, y sus precursores
BR112018077390A2 (pt) 2016-06-29 2019-04-09 Kezar Life Sciences sais cristalinos de peptídeo epoxicentona inibidor de imunoproteassoma
WO2018044991A1 (en) 2016-08-30 2018-03-08 Dow Agrosciences Llc Thiopicolinamide compounds with fungicidal activity
WO2018044996A1 (en) 2016-08-30 2018-03-08 Dow Agrosciences Llc Picolinamides as fungicides
US10111432B2 (en) 2016-08-30 2018-10-30 Dow Agrosciences Llc Picolinamide N-oxide compounds with fungicidal activity
WO2018045010A1 (en) 2016-08-30 2018-03-08 Dow Agrosciences Llc Pyrido-1,3-oxazine-2,4-dione compounds with fungicidal activity
WO2018057453A1 (en) 2016-09-21 2018-03-29 Amgen Inc. Immediate release formulations for oprozomib
US20180161279A1 (en) 2016-12-14 2018-06-14 Amgen Inc. Gastro-retentive modified release dosage forms for oprozomib and process to make thereof
BR102018000183B1 (pt) 2017-01-05 2023-04-25 Dow Agrosciences Llc Picolinamidas, composição para controle de um patógeno fúngico, e método para controle e prevenção de um ataque por fungos em uma planta
TWI774761B (zh) 2017-05-02 2022-08-21 美商科迪華農業科技有限責任公司 用於穀物中的真菌防治之協同性混合物
WO2018204438A1 (en) 2017-05-02 2018-11-08 Dow Agrosciences Llc Use of an acyclic picolinamide compound as a fungicide for fungal diseases on turfgrasses
TW201842851A (zh) 2017-05-02 2018-12-16 美商陶氏農業科學公司 用於穀類中的真菌防治之協同性混合物
BR112020005079A2 (pt) 2017-09-14 2020-09-15 Glaxosmithkline Intellectual Property Development Limited tratamento de combinação para câncer
BR102019004480B1 (pt) 2018-03-08 2023-03-28 Dow Agrosciences Llc Picolinamidas como fungicidas
EP3866597A4 (en) 2018-10-15 2022-06-29 Corteva Agriscience LLC Methods for sythesis of oxypicolinamides

Family Cites Families (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4733665C2 (en) 1985-11-07 2002-01-29 Expandable Grafts Partnership Expandable intraluminal graft and method and apparatus for implanting an expandable intraluminal graft
US5135919A (en) 1988-01-19 1992-08-04 Children's Medical Center Corporation Method and a pharmaceutical composition for the inhibition of angiogenesis
US5441944A (en) 1989-04-23 1995-08-15 The Trustees Of The University Of Pennsylvania Substituted cyclodextrin sulfates and their uses as growth modulating agents
US4990448A (en) 1989-08-04 1991-02-05 Bristol-Myers Company Bu-4061T
US5071957A (en) 1989-08-04 1991-12-10 Bristol-Myers Company Antibiotic BU-4061T
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
KR0166088B1 (ko) 1990-01-23 1999-01-15 . 수용해도가 증가된 시클로덱스트린 유도체 및 이의 용도
EP0518955A4 (en) 1990-03-05 1993-09-22 Cephalon, Inc. Chymotrypsin-like proteases and their inhibitors
US5561134A (en) * 1990-09-25 1996-10-01 Rhone-Poulenc Rorer Pharmaceuticals Inc. Compounds having antihypertensive, cardioprotective, anti-ischemic and antilipolytic properties
US5340736A (en) 1991-05-13 1994-08-23 The President & Fellows Of Harvard College ATP-dependent protease and use of inhibitors for same in the treatment of cachexia and muscle wasting
EP0955365A3 (en) * 1992-06-09 2000-12-20 Chiron Corporation Crystallization of M-CSF
GB9300048D0 (en) 1993-01-04 1993-03-03 Wellcome Found Endothelin converting enzyme inhibitors
TW380137B (en) 1994-03-04 2000-01-21 Merck & Co Inc Process for making an epoxide
US5693617A (en) 1994-03-15 1997-12-02 Proscript, Inc. Inhibitors of the 26s proteolytic complex and the 20s proteasome contained therein
US6660268B1 (en) 1994-03-18 2003-12-09 The President And Fellows Of Harvard College Proteasome regulation of NF-KB activity
US6506876B1 (en) 1994-10-11 2003-01-14 G.D. Searle & Co. LTA4 hydrolase inhibitor pharmaceutical compositions and methods of use
US6083903A (en) 1994-10-28 2000-07-04 Leukosite, Inc. Boronic ester and acid compounds, synthesis and uses
DE19505263A1 (de) 1995-02-16 1996-08-22 Consortium Elektrochem Ind Verfahren zur Reinigung von wasserlöslichen Cyclodextrinderivaten
US6335358B1 (en) 1995-04-12 2002-01-01 President And Fellows Of Harvard College Lactacystin analogs
US6150415A (en) 1996-08-13 2000-11-21 The Regents Of The University Of California Epoxide hydrolase complexes and methods therewith
AU4499697A (en) 1996-09-13 1998-04-02 New York University Method for treating parasitic diseases with proteasome inhibitors
SI0932617T1 (en) 1996-10-18 2002-06-30 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases, particularly hepatitis c virus ns3 protease
JP4080541B2 (ja) 1996-10-18 2008-04-23 バーテックス ファーマシューティカルズ インコーポレイテッド セリンプロテアーゼ、特にc型肝炎ウイルスns3プロテアーゼの阻害因子
US6046177A (en) 1997-05-05 2000-04-04 Cydex, Inc. Sulfoalkyl ether cyclodextrin based controlled release solid pharmaceutical formulations
US5874418A (en) 1997-05-05 1999-02-23 Cydex, Inc. Sulfoalkyl ether cyclodextrin based solid pharmaceutical formulations and their use
AU750207B2 (en) 1997-06-13 2002-07-11 Cydex Pharmaceuticals, Inc. Polar drug or prodrug compositions with extended shelf-life storage and a method of making thereof
US6133308A (en) 1997-08-15 2000-10-17 Millennium Pharmaceuticals, Inc. Synthesis of clasto-lactacystin beta-lactone and analogs thereof
US6100282A (en) * 1998-01-02 2000-08-08 Hoffman-La Roche Inc. Thiazole derivatives
US6075150A (en) 1998-01-26 2000-06-13 Cv Therapeutics, Inc. α-ketoamide inhibitors of 20S proteasome
US6099851A (en) 1998-06-02 2000-08-08 Weisman; Kenneth M. Therapeutic uses of leuprolide acetate
US6838436B1 (en) 1998-07-10 2005-01-04 Osteoscreen Inc. Inhibitors of proteasomal activity for stimulating bone growth
US6462019B1 (en) 1998-07-10 2002-10-08 Osteoscreen, Inc. Inhibitors of proteasomal activity and production for stimulating bone growth
US6902721B1 (en) 1998-07-10 2005-06-07 Osteoscreen, Inc. Inhibitors of proteasomal activity for stimulating bone growth
US6204257B1 (en) 1998-08-07 2001-03-20 Universtiy Of Kansas Water soluble prodrugs of hindered alcohols
KR20010080267A (ko) 1998-10-20 2001-08-22 밀레니엄 파머슈티컬스 인코퍼레이티드 프로테아솜 억제제 약물 작용을 모니터링하는 방법
US6492333B1 (en) 1999-04-09 2002-12-10 Osteoscreen, Inc. Treatment of myeloma bone disease with proteasomal and NF-κB activity inhibitors
US6831099B1 (en) 1999-05-12 2004-12-14 Yale University Enzyme inhibition
US6114365A (en) * 1999-08-12 2000-09-05 Pharmacia & Upjohn S.P.A. Arylmethyl-carbonylamino-thiazole derivatives, process for their preparation, and their use as antitumor agents
JP2003528039A (ja) 1999-10-20 2003-09-24 オステオスクリーン,インコーポレイテッド 骨および毛成長を刺激するためのプロテアソーム活性のインヒビター
DK1326632T3 (da) 2000-10-12 2007-01-15 Viromics Gmbh Protease-inhibitorer til behandling af infektioner af hepatitisvirus
GB0114185D0 (en) * 2001-06-12 2001-08-01 Protherics Molecular Design Lt Compounds
AU2002243646B2 (en) 2001-01-25 2006-06-22 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Formulation of boronic acid compounds
PT1392355E (pt) 2001-05-21 2007-03-30 Alcon Inc Utilização de inibidores do proteassoma para tratar distúrbios associados à secura ocular
US7524883B2 (en) 2002-01-08 2009-04-28 Eisai R&D Management Co., Ltd. Eponemycin and epoxomicin analogs and uses thereof
US20040116329A1 (en) 2002-01-29 2004-06-17 Epstein Stephen E. Inhibition of proteasomes to prevent restenosis
WO2003086283A2 (en) 2002-04-09 2003-10-23 Greenville Hospital System Metastasis modulating activity of highly sulfated oligosaccharides
US7968569B2 (en) 2002-05-17 2011-06-28 Celgene Corporation Methods for treatment of multiple myeloma using 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione
US7902185B2 (en) 2002-06-03 2011-03-08 Als Therapy Development Foundation, Inc. Treatment of neurodegenerative diseases using proteasome modulators
US20030224469A1 (en) 2002-06-03 2003-12-04 Buchholz Tonia J. Methods and kits for assays utilizing fluorescence polarization
AU2003256847A1 (en) 2002-07-26 2004-02-16 Advanced Research And Technology Institute At Indiana University Method of treating cancer
US7189740B2 (en) 2002-10-15 2007-03-13 Celgene Corporation Methods of using 3-(4-amino-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione for the treatment and management of myelodysplastic syndromes
TW200418791A (en) 2003-01-23 2004-10-01 Bristol Myers Squibb Co Pharmaceutical compositions for inhibiting proteasome
HUE028247T2 (en) 2003-04-08 2016-12-28 Novartis Ag A solid oral composition comprising S1P receptor agonist and sugar alcohol
US8338180B2 (en) 2003-06-10 2012-12-25 The J. David Gladstone Institutes Methods for treating lentivirus infections
US7012063B2 (en) 2003-06-13 2006-03-14 Children's Medical Center Corporation Reducing axon degeneration with proteasome inhibitors
EP1729724A4 (en) 2003-12-31 2008-07-23 Cydex Inc INHALATION FORMULATION WITH SULFOALKYL ETHER GAMMA CYCLODEXTRIN AND CORTICOSTEROID
GB0400804D0 (en) 2004-01-14 2004-02-18 Innoscience Technology Bv Pharmaceutical compositions
US20050228031A1 (en) * 2004-04-13 2005-10-13 Bilodeau Mark T Tyrosine kinase inhibitors
US8198270B2 (en) * 2004-04-15 2012-06-12 Onyx Therapeutics, Inc. Compounds for proteasome enzyme inhibition
US7232818B2 (en) * 2004-04-15 2007-06-19 Proteolix, Inc. Compounds for enzyme inhibition
SG152239A1 (en) 2004-04-15 2009-05-29 Proteolix Inc Compounds for proteasome enzyme inhibition
US20050256324A1 (en) 2004-05-10 2005-11-17 Proteolix, Inc. Synthesis of amino acid keto-epoxides
PT2030981E (pt) 2004-05-10 2014-10-14 Onyx Therapeutics Inc Compostos para inibição da enzima proteassoma
CN101044157B (zh) * 2004-08-06 2011-07-13 普罗特奥里克斯公司 用于抑制蛋白酶体的化合物
US20060088471A1 (en) 2004-10-20 2006-04-27 Proteolix, Inc. Compounds for enzyme inhibition
JP4990155B2 (ja) 2004-12-07 2012-08-01 プロテオリックス, インコーポレイテッド プロテアソームを阻害するための組成物
US7468383B2 (en) 2005-02-11 2008-12-23 Cephalon, Inc. Proteasome inhibitors and methods of using the same
EP1863513A2 (en) 2005-03-11 2007-12-12 The University of North Carolina at Chapel Hill Potent and specific immunoproteasome inhibitors
ATE553198T1 (de) 2005-04-15 2012-04-15 Geron Corp Behandlung von krebs durch die kombinierte hemmung der proteasom- und telomeraseaktivitäten
GT200600350A (es) 2005-08-09 2007-03-28 Formulaciones líquidas
CN105237621A (zh) * 2005-11-09 2016-01-13 欧尼斯治疗公司 用于酶抑制的化合物
AR057227A1 (es) 2005-12-09 2007-11-21 Centocor Inc Metodo para usar antagonistas de il6 con inhibidores del proteasoma
US20070207950A1 (en) 2005-12-21 2007-09-06 Duke University Methods and compositions for regulating HDAC6 activity
WO2007122686A1 (ja) 2006-04-14 2007-11-01 Eisai R & D Management Co., Ltd. ベンズイミダゾール化合物
DE102006026464A1 (de) 2006-06-01 2007-12-06 Virologik Gmbh Innovationszentrum Medizintechnik Und Pharma Pharmazeutische Zusammensetzung zur Behandlung von Virusinfektionen und / oder Tumorerkrankungen durch Inhibition der Proteinfaltung und des Proteinabbaus
EP2041158B1 (en) * 2006-06-19 2013-04-17 Onyx Therapeutics, Inc. Peptide epoxyketones for proteasome inhibition
WO2008033807A2 (en) 2006-09-13 2008-03-20 The Arizona Board Of Regents On Behalf Of The University Of Arizona Synergistic combinations of antineoplastic thiol-binding mitochondrial oxidants and antineoplastic proteasome inhibitors for the treatment of cancer
US8601054B2 (en) * 2006-12-07 2013-12-03 International Business Machines Corporation Project-related communications
AU2008209555A1 (en) 2007-01-23 2008-07-31 Gloucester Pharmaceuticals, Inc. Combination therapy comprising romidepsin and I.A. bortezomib
WO2008140782A2 (en) * 2007-05-10 2008-11-20 Proteolix, Inc. Compounds for enzyme inhibition
US7442830B1 (en) 2007-08-06 2008-10-28 Millenium Pharmaceuticals, Inc. Proteasome inhibitors
WO2009020448A1 (en) 2007-08-06 2009-02-12 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
CL2008002966A1 (es) * 2007-10-04 2010-06-25 Onyx Therapeutics Inc Compuesto tetrapeptido ceto-epoxido cristalino; sal citrato cristalina del compuesto; metodos de preparacion; compuesto intermediario cristalino; metodo de preparacion; y uso para tratar cancer, enfermedad autoinmune, afeccion relacionada con trasplante, enfermedad neurodegenerativa, afeccion asociada con fibrosis, entre otros.
WO2009051581A1 (en) 2007-10-16 2009-04-23 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
US7838673B2 (en) 2007-10-16 2010-11-23 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
US20090131367A1 (en) 2007-11-19 2009-05-21 The Regents Of The University Of Colorado Combinations of HDAC Inhibitors and Proteasome Inhibitors
MX349769B (es) 2008-06-17 2017-08-11 Millennium Pharm Inc Compuestos de éster boronato y composiciones farmacéuticas de los mismos.
AR075090A1 (es) 2008-09-29 2011-03-09 Millennium Pharm Inc Derivados de acido 1-amino-2-ciclobutiletilboronico inhibidores de proteosoma,utiles como agentes anticancerigenos, y composiciones farmaceuticas que los comprenden.
EA035100B1 (ru) 2008-10-21 2020-04-28 Оникс Терапьютикс, Инк. Комбинированная терапия с применением пептид эпоксикетонов
TWI504598B (zh) 2009-03-20 2015-10-21 Onyx Therapeutics Inc 結晶性三肽環氧酮蛋白酶抑制劑
CN101928329B (zh) 2009-06-19 2013-07-17 北京大学 三肽硼酸(酯)类化合物、其制备方法和应用
WO2011060179A1 (en) 2009-11-13 2011-05-19 Onyx Therapeutics, Inc Use of peptide epoxyketones for metastasis suppression
US9359398B2 (en) 2010-03-01 2016-06-07 Onyx Therapeutics, Inc. Compounds for immunoproteasome inhibition
EA029521B1 (ru) 2010-03-31 2018-04-30 Милленниум Фармасьютикалз, Инк. Производные 1-амино-2-циклопропилэтилбороновой кислоты
BR112012025264A2 (pt) 2010-04-07 2019-09-24 Onyx Therapeutics Inc inibidor de imunoproteassoma de e´poxicetona peptídica cristalina.

Also Published As

Publication number Publication date
RS53746B1 (en) 2015-06-30
US8604215B2 (en) 2013-12-10
KR20110132577A (ko) 2011-12-08
PL2408758T3 (pl) 2015-03-31
US20120077855A1 (en) 2012-03-29
IL215174A (en) 2016-03-31
HRP20161654T1 (hr) 2017-01-27
SMT201700068B (it) 2017-03-08
PE20120645A1 (es) 2012-06-07
HRP20150014T1 (hr) 2015-02-27
HK1204952A1 (en) 2015-12-11
PT2813241T (pt) 2016-12-23
HUE032430T2 (en) 2017-09-28
BRPI1009369A2 (pt) 2016-10-11
EP2408758A1 (en) 2012-01-25
EP2813241A1 (en) 2014-12-17
EA024672B1 (ru) 2016-10-31
CO6430433A2 (es) 2012-04-30
ZA201601686B (en) 2016-11-30
SG2014011373A (en) 2014-05-29
JP2012521363A (ja) 2012-09-13
SI2813241T1 (sl) 2017-03-31
HK1162476A1 (en) 2012-08-31
EP2408758A4 (en) 2012-09-05
MX2011009777A (es) 2011-10-17
NI201100170A (es) 2012-02-21
GEP20156392B (en) 2015-11-10
ES2614557T3 (es) 2017-05-31
ZA201106826B (en) 2018-11-28
SG174446A1 (en) 2011-10-28
US20120088903A1 (en) 2012-04-12
CN102428075B (zh) 2016-05-11
JP5723357B2 (ja) 2015-05-27
LT2813241T (lt) 2017-01-10
WO2010108172A1 (en) 2010-09-23
TN2011000470A1 (en) 2013-03-27
HN2011002459A (es) 2015-03-16
US20150175656A1 (en) 2015-06-25
AP2013007137A0 (en) 2013-09-30
CR20110491A (es) 2012-01-19
US20100240903A1 (en) 2010-09-23
EP2813241B1 (en) 2016-11-16
ES2527619T3 (es) 2015-01-27
IL215174A0 (en) 2011-12-29
AR075899A1 (es) 2011-05-04
TWI504598B (zh) 2015-10-21
SMT201500009B (it) 2015-03-05
AP3513A (en) 2016-01-05
US9403868B2 (en) 2016-08-02
PL2813241T3 (pl) 2017-04-28
CL2011002326A1 (es) 2012-03-23
CU20110176A7 (es) 2012-04-15
TW201043623A (en) 2010-12-16
NZ618432A (en) 2015-04-24
SA114350283B1 (ar) 2016-06-08
CA2755971A1 (en) 2010-09-23
US20140024804A1 (en) 2014-01-23
CN102428075A (zh) 2012-04-25
MA33197B1 (fr) 2012-04-02
AU2010226410A1 (en) 2011-11-03
AU2010226410B2 (en) 2015-09-17
SI2408758T1 (sl) 2015-01-30
ECSP11011341A (es) 2011-10-31
CA2755971C (en) 2017-05-16
EA201300860A1 (ru) 2014-04-30
MY156522A (en) 2016-02-26
EA201171151A1 (ru) 2012-04-30
EA020973B1 (ru) 2015-03-31
DOP2011000286A (es) 2011-10-15
DK2813241T3 (en) 2017-01-16
MX343562B (es) 2016-11-09
US8822512B2 (en) 2014-09-02
KR101729344B1 (ko) 2017-04-21
NZ595847A (en) 2013-12-20
RS55431B1 (sr) 2017-04-28
CY1118359T1 (el) 2017-06-28
US9051353B2 (en) 2015-06-09
ME01277B (me) 2010-03-22
SA110310221B1 (ar) 2014-04-28
DK2408758T3 (da) 2014-12-01
PT2408758E (pt) 2014-11-04

Similar Documents

Publication Publication Date Title
EP2408758B1 (en) Crystalline tripeptide epoxy ketone protease inhibitors
CA2701778C (en) Crystalline peptide epoxy ketone protease inhibitors and the synthesis of amino acid keto-epoxides
AU2013203857B2 (en) Crystalline tripeptide epoxy ketone protease inhibitors
AU2013203566B2 (en) Crystalline peptide epoxy ketone protease inhibitors and the synthesis of amino acid keto-epoxides

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20111018

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

AX Request for extension of the european patent

Extension state: AL BA RS

RAX Requested extension states of the european patent have changed

Extension state: BA

Payment date: 20111018

Extension state: AL

Payment date: 20111018

Extension state: RS

Payment date: 20111018

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602010019713

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: C07D0277560000

Ipc: A61K0038550000

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1162476

Country of ref document: HK

A4 Supplementary search report drawn up and despatched

Effective date: 20120803

RIC1 Information provided on ipc code assigned before grant

Ipc: C07D 417/12 20060101ALI20120730BHEP

Ipc: C07K 5/08 20060101ALI20120730BHEP

Ipc: A61K 38/55 20060101AFI20120730BHEP

17Q First examination report despatched

Effective date: 20130626

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20140521

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

AX Request for extension of the european patent

Extension state: AL BA RS

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

Ref country code: CH

Ref legal event code: NV

Representative=s name: NOVAGRAAF INTERNATIONAL SA, CH

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20141027

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 692283

Country of ref document: AT

Kind code of ref document: T

Effective date: 20141115

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20141126

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602010019713

Country of ref document: DE

Effective date: 20141204

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: HR

Ref legal event code: TUEP

Ref document number: P20150014

Country of ref document: HR

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2527619

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20150127

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20141022

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E009991

Country of ref document: EE

Effective date: 20141209

REG Reference to a national code

Ref country code: HR

Ref legal event code: T1PR

Ref document number: P20150014

Country of ref document: HR

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E022038

Country of ref document: HU

REG Reference to a national code

Ref country code: PL

Ref legal event code: T3

REG Reference to a national code

Ref country code: SK

Ref legal event code: T3

Ref document number: E 17788

Country of ref document: SK

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20150400029

Country of ref document: GR

Effective date: 20150220

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602010019713

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1162476

Country of ref document: HK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20150723

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 692283

Country of ref document: AT

Kind code of ref document: T

Effective date: 20141022

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 7

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 9

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20150014

Country of ref document: HR

Payment date: 20190218

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20190312

Year of fee payment: 10

Ref country code: NO

Payment date: 20190308

Year of fee payment: 10

Ref country code: PL

Payment date: 20190212

Year of fee payment: 10

Ref country code: BG

Payment date: 20190226

Year of fee payment: 10

Ref country code: CH

Payment date: 20190314

Year of fee payment: 10

Ref country code: FI

Payment date: 20190311

Year of fee payment: 10

Ref country code: GB

Payment date: 20190320

Year of fee payment: 10

Ref country code: CZ

Payment date: 20190228

Year of fee payment: 10

Ref country code: LU

Payment date: 20190308

Year of fee payment: 10

Ref country code: MC

Payment date: 20190227

Year of fee payment: 10

Ref country code: IS

Payment date: 20190207

Year of fee payment: 10

Ref country code: IT

Payment date: 20190326

Year of fee payment: 10

Ref country code: IE

Payment date: 20190311

Year of fee payment: 10

Ref country code: LT

Payment date: 20190221

Year of fee payment: 10

Ref country code: RO

Payment date: 20190301

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: AT

Payment date: 20190226

Year of fee payment: 10

Ref country code: DK

Payment date: 20190312

Year of fee payment: 10

Ref country code: HR

Payment date: 20190218

Year of fee payment: 10

Ref country code: EE

Payment date: 20190228

Year of fee payment: 10

Ref country code: HU

Payment date: 20190221

Year of fee payment: 10

Ref country code: TR

Payment date: 20190312

Year of fee payment: 10

Ref country code: GR

Payment date: 20190213

Year of fee payment: 10

Ref country code: SI

Payment date: 20190228

Year of fee payment: 10

Ref country code: BE

Payment date: 20190215

Year of fee payment: 10

Ref country code: FR

Payment date: 20190213

Year of fee payment: 10

Ref country code: LV

Payment date: 20190305

Year of fee payment: 10

Ref country code: SM

Payment date: 20190227

Year of fee payment: 10

Ref country code: SE

Payment date: 20190311

Year of fee payment: 10

Ref country code: NL

Payment date: 20190313

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SK

Payment date: 20190218

Year of fee payment: 10

Ref country code: MK

Payment date: 20190304

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20190401

Year of fee payment: 10

Ref country code: PT

Payment date: 20190321

Year of fee payment: 10

Ref country code: CY

Payment date: 20190318

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MT

Payment date: 20190315

Year of fee payment: 10

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602010019713

Country of ref document: DE

REG Reference to a national code

Ref country code: HR

Ref legal event code: PBON

Ref document number: P20150014

Country of ref document: HR

Effective date: 20200322

Ref country code: FI

Ref legal event code: MAE

REG Reference to a national code

Ref country code: EE

Ref legal event code: MM4A

Ref document number: E009991

Country of ref document: EE

Effective date: 20200331

REG Reference to a national code

Ref country code: NO

Ref legal event code: MMEP

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

Ref country code: PT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200922

Ref country code: CY

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

Ref country code: CZ

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

Ref country code: FI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

Ref country code: RO

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20200331

REG Reference to a national code

Ref country code: NL

Ref legal event code: MM

Effective date: 20200401

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 692283

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200322

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

REG Reference to a national code

Ref country code: SK

Ref legal event code: MM4A

Ref document number: E 17788

Country of ref document: SK

Effective date: 20200322

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20200331

REG Reference to a national code

Ref country code: LT

Ref legal event code: MM4D

Effective date: 20200322

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200401

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

REG Reference to a national code

Ref country code: SI

Ref legal event code: KO00

Effective date: 20201113

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

Ref country code: GR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201008

Ref country code: NO

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201001

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

Ref country code: HR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200323

Ref country code: LT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

Ref country code: BG

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201130

Ref country code: SM

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201008

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

Ref country code: HU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200323

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

Ref country code: SI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200323

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20200322

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20210806

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200323

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200322